Metabolic Regulation of Ferroptosis in Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Metabolic Regulation of Ferroptosis in Cancer biology Review Metabolic Regulation of Ferroptosis in Cancer Min Ji Kim 1,2,† , Greg Jiho Yun 1,2,† and Sung Eun Kim 1,2,* 1 Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 305-350, Korea; [email protected] (M.J.K.); [email protected] (G.J.Y.) 2 Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 305-350, Korea * Correspondence: [email protected]; Tel.: +82-2-3290-5647 † These authors contributed equally to this work as co-first authors. Simple Summary: Ferroptosis is a recently defined nonapoptotic form of cell death that is associated with various human diseases, including cancer. As ferroptosis is caused by an overdose of lipid peroxidation resulting from dysregulation of the cellular antioxidant system, it is inherently closely associated with cellular metabolism. Here, we provide an updated review of the recent studies that have shown mechanisms of metabolic regulation of ferroptosis in the context of cancer. Abstract: Ferroptosis is a unique cell death mechanism that is executed by the excessive accumulation of lipid peroxidation in cells. The relevance of ferroptosis in multiple human diseases such as neurodegeneration, organ damage, and cancer is becoming increasingly evident. As ferroptosis is deeply intertwined with metabolic pathways such as iron, cyst(e)ine, glutathione, and lipid metabolism, a better understanding of how ferroptosis is regulated by these pathways will enable the precise utilization or prevention of ferroptosis for therapeutic uses. In this review, we present an update of the mechanisms underlying diverse metabolic pathways that can regulate ferroptosis in cancer. Keywords: ferroptosis; iron metabolism; cyst(e)ine metabolism; SLC7A11; glutathione metabolism; Citation: Kim, M.J.; Yun, G.J.; Kim, GPX4; lipid peroxidation; reactive oxygen species S.E. Metabolic Regulation of Ferroptosis in Cancer. Biology 2021, 10, 83. https://doi.org/10.3390/ biology10020083 1. Introduction Academic Editor: Annalisa Pinsino “Every true story ends in death” are the words of Ernest Hemingway. Cell death is an Received: 29 December 2020 important aspect of mammalian development and homeostasis and is tightly integrated Accepted: 20 January 2021 with the physiological and pathological state of an organism. The proper orchestration Published: 22 January 2021 of cell death both spatially and temporally is critical for development and, consequently, the malfunction of cell death mechanisms can contribute to various human diseases. Publisher’s Note: MDPI stays neutral Ferroptosis is a nonapoptotic cell death mechanism characterized by iron-dependent ac- with regard to jurisdictional claims in cumulation of lipid peroxides. Importantly, ferroptosis has been implicated in neurode- published maps and institutional affil- generative diseases, kidney degeneration, and ischemic injury in multiple organs. Studies iations. using ferroptosis inhibitors have shown to be effective in ischemia/reperfusion-induced damage and in models of Huntington’s disease and Parkinson’s disease, suggesting that ferroptosis is a promising therapeutic target for the treatment of these diseases. On the other hand, the possibility of harnessing ferroptosis as a method for cancer therapy is also Copyright: © 2021 by the authors. gaining much attraction. Indeed, the original study that led to the identification of erastin, Licensee MDPI, Basel, Switzerland. a ferroptosis-inducing drug, was aimed for the selective killing of RAS-mutant cancer This article is an open access article cells [1]. Since then, multiple lines of research have suggested the important role of ferrop- distributed under the terms and tosis in cancer development and treatment. For example, ferroptosis has been suggested conditions of the Creative Commons to be a critical factor for the activities of the tumor suppressors p53, BRCA1-associated Attribution (CC BY) license (https:// protein 1 (BAP1), and fumarase [2–4]. Ferroptosis can also play a tumor-suppressive role creativecommons.org/licenses/by/ during the metastasis of cancer cells as melanoma cells in the lymph are protected from 4.0/). Biology 2021, 10, 83. https://doi.org/10.3390/biology10020083 https://www.mdpi.com/journal/biology Biology 2021, 10, 83 2 of 21 ferroptosis and, thus, can form more metastases than those in the blood [5]. Similarly, circulating tumor cells from melanoma patients have been shown to regulate lipogenesis and iron homeostasis pathways to confer resistance to ferroptosis [6]. Several types of cancer cells are susceptible to ferroptosis; thus, ferroptosis may rep- resent a novel mode of anticancer therapy. Cancer types that show high sensitivity to ferroptosis include renal cell carcinoma, diffuse large B cell lymphoma, adrenocortical carcinoma, and ovarian cancer [7–9]. Recently, it was reported that therapy-resistant or drug-tolerant cells, which are represented by a high mesenchymal state, depend on the glutathione peroxidase 4 (GPX4) pathway to evade ferroptosis, suggesting that ferroptosis induction represents an anticancer strategy for these therapy resistant cancer cells [10,11]. Similarly, platinum-tolerant or cisplatin-resistant cancer cells were also shown to exhibit increased vulnerability to ferroptosis [12,13]. This was also shown in the case of melanoma, where the dedifferentiation state of these cells was shown to correlate with increased sensi- tivity to ferroptosis [14]. The function of ferroptosis in cancer can also involve multiple cell types in the tumor microenvironment. A recent study shows that neutrophils can induce ferroptosis in glioblastoma cells by transferring myeloperoxidase-containing granules into the cancer cells [15]. Additionally, immunotherapy activated CD8+ T cells can induce fer- roptosis in cancer cells by interferon gamma-mediated downregulation of the two subunits − of system Xc , solute carrier family 7 member 11 (SLC7A11) and SLC3A2, leading to a decrease in the antioxidant capacity of cancer cells [16]. This effect can also synergize with radiotherapy through independent SLC7A11 suppression by the ataxia-telangiectasia mutated (ATM) serine/threonine kinase [17]. For the induction of ferroptosis in cancer, agents that inhibit cystine uptake via the − cystine/glutamate antiporter, system Xc , such as sulfasalazine, can induce ferropto- sis and arrest tumor growth [18]. The multikinase inhibitor sorafenib, which is used for advanced hepatocellular carcinoma, has also been shown to induce ferroptosis by − inhibiting system Xc [19]. Similarly, the systemic depletion of cyst(e)ine using an engi- neered cyst(e)ine-degrading enzyme conjugate can trigger ferroptosis and arrest tumor growth [20] and the antimalarial drug, artesunate, has also been identified as an activa- tor of ferroptosis [21]. Furthermore, ferroptosis can synergize with cisplatin to increase cytotoxicity in cancer cells [22] and radiation therapy can also synergize with ferroptosis inducers [23,24]. Although several ferroptosis-inducing agents exist, including erastin, RSL3, and ML210, further studies to enhance the pharmacokinetic properties are needed for use in clinical settings [7,25]. In essence, ferroptosis is a process that occurs through metabolic dysregulation. Ferroptosis is modulated by perturbation of lipid repair systems involving glutathione and GPX4, the enzyme that converts toxic lipid hydroperoxides to nontoxic lipid alcohols. The term ferroptosis was coined in 2012, describing the cell death induced by erastin, that inhibits the import of cystine and leads to the depletion of glutathione and inactivation of GPX4 [26]. Inactivation of GPX4 through the depletion of glutathione or through direct GPX4 inhibition results in the accumulation of lipid peroxidation that leads to cell death. Ferroptosis can be suppressed by iron chelators, lipophilic antioxidants, lipid peroxidation inhibitors, or the depletion of polyunsaturated fatty acids (PUFAs). Thus, ferroptosis is characterized by the perturbation of an intricate metabolic network which we will describe in detail in the following sections. 2. Iron Metabolism As the name ‘ferroptosis’ infers, the requirement of intracellular iron is a fundamental property of ferroptosis. As an important trace element in the human body, iron regulates numerous biological processes and, therefore, dysregulation of iron content or distribution can result in intracellular iron accumulation, which can lead to the damage of cells, tissues, and organs. Elaborate regulatory systems have evolved to maintain iron levels at sufficient yet safe concentrations in cells. Such homeostasis of iron metabolism is achieved by the coordination of iron uptake, utilization, recycling, storage, and export [27]. The iron Biology 2021, 10, x 3 of 21 Biology 2021, 10, 83 3 of 21 [27]. The iron dependency of ferroptosis was initially demonstrated by the use of iron chelatorsdependency to inhibit of ferroptosis ferroptosis. was initially Additionally, demonstrated in the same by the study, use of iron iron responsive chelators to element inhibit bindingferroptosis. protein Additionally, 2 (IRP2), a inmaster the same regulator study, of iron iron responsive metabolism, element was found binding in an protein erastin- 2 (IRP2),resistance a master screen regulatorestablishing of ironthat ferroptosis metabolism, is wasintricately found modulated in an erastin-resistance by iron metabolism. screen Perturbationestablishing thatof iron ferroptosis metabolism is intricately
Recommended publications
  • Dysregulated Hepatic Methionine
    Virginia Commonwealth University VCU Scholars Compass Internal Medicine Publications Dept. of Internal Medicine 2015 Dysregulated Hepatic Methionine Metabolism Drives Homocysteine Elevation in Diet-Induced Nonalcoholic Fatty Liver Disease Tommy Pacana Virginia Commonwealth University, [email protected] Sophie Cazanave Virginia Commonwealth University Aurora Verdianelli Virginia Commonwealth University See next page for additional authors Follow this and additional works at: http://scholarscompass.vcu.edu/intmed_pubs Part of the Medicine and Health Sciences Commons Copyright: © 2015 Pacana et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited Downloaded from http://scholarscompass.vcu.edu/intmed_pubs/98 This Article is brought to you for free and open access by the Dept. of Internal Medicine at VCU Scholars Compass. It has been accepted for inclusion in Internal Medicine Publications by an authorized administrator of VCU Scholars Compass. For more information, please contact [email protected]. Authors Tommy Pacana, Sophie Cazanave, Aurora Verdianelli, Viashali Patel, Hae-Ki Min, Faridoddin Mirshahi, Eoin Quinlavin, and Arun J. Sanyal This article is available at VCU Scholars Compass: http://scholarscompass.vcu.edu/intmed_pubs/98 RESEARCH ARTICLE Dysregulated Hepatic Methionine Metabolism Drives Homocysteine Elevation in Diet-Induced Nonalcoholic Fatty
    [Show full text]
  • The Role of Glutathionperoxidase 4 (GPX4) in Hematopoiesis and Leukemia
    The role of glutathionperoxidase 4 (GPX4) in hematopoiesis and leukemia von Kira Célénie Stahnke Inaugral- Dissertation zur Erlangung der Doktorwürde der TierärztliChen Fakultät der Ludwig-Maximilians-Universität MünChen The role of glutathionperoxidase 4 (GPX4) in hematopoiesis and leukemia von Kira Célénie Stahnke aus Dortmund München 2016 Aus dem VeterinärwissensChaftliChen Department der TierärztliChen Fakultät der Ludwig-Maximilians-Universität München Lehrstuhl für Molekulare Tierzucht und Biotechnologie Arbeit angefertigt unter der Leitung von Univ.-Prof. Dr. Eckhard Wolf Angefertigt am Institut für Experimentelle TumorforsChung Universitätsklinikum Ulm Mentor: Prof. Dr. Christian Buske GedruCkt mit Genehmigung der TierärztliChen Fakultät der Ludwig-Maximilians-Universität München Dekan: Univ.-Prof. Dr. JoaChim Braun Berichterstatter: Univ.-Prof. Dr. Eckard Wolf Korreferent/en: Priv.-Doz. Dr. Bianka Schulz Tag der Promotion: 06.02.2016 Table of contents List of Abbreviations..............................................................................2 1 Introduction ........................................................................................6 1.1 ROS and oxidative stress in physiology and pathology..................................................... 6 1.1.1 SourCes of Reactive Oxygen SpeCies..................................................................................................6 1.1.2 PhysiologiCal role of ROS........................................................................................................................8
    [Show full text]
  • Review Article Cystathionine -Synthase in Physiology and Cancer
    Hindawi BioMed Research International Volume 2018, Article ID 3205125, 11 pages https://doi.org/10.1155/2018/3205125 Review Article Cystathionine �-Synthase in Physiology and Cancer Haoran Zhu,1,2 Shaun Blake,1,2 Keefe T. Chan,1 Richard B. Pearson ,1,2,3,4 and Jian Kang 1 1 Division of Research, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia 2Sir Peter MacCallum Department of Oncology, Australia 3Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia 4Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3168, Australia Correspondence should be addressed to Richard B. Pearson; [email protected] Received 23 March 2018; Accepted 29 May 2018; Published 28 June 2018 Academic Editor: Maria L. Tornesello Copyright © 2018 Haoran Zhu et al. Tis is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Cystathionine �-synthase (CBS) regulates homocysteine metabolism and contributes to hydrogen sulfde (H2S) biosynthesis through which it plays multifunctional roles in the regulation of cellular energetics, redox status, DNA methylation, and protein modifcation. Inactivating mutations in CBS contribute to the pathogenesis of the autosomal recessive disease CBS-defcient homocystinuria. Recent studies demonstrating that CBS promotes colon and ovarian cancer growth in preclinical models highlight a newly identifed oncogenic role for CBS. On the contrary, tumor-suppressive efects of CBS have been reported in other cancer types, suggesting context-dependent roles of CBS in tumor growth and progression. Here, we review the physiological functions of CBS, summarize the complexities regarding CBS research in oncology, and discuss the potential of CBS and its key metabolites, including homocysteine and H2S, as potential biomarkers for cancer diagnosis or therapeutic targets for cancer treatment.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Anti-AHCY / Adenosylhomocysteinase Antibody (ARG57333)
    Product datasheet [email protected] ARG57333 Package: 100 μl anti-AHCY / Adenosylhomocysteinase antibody Store at: -20°C Summary Product Description Rabbit Polyclonal antibody recognizes AHCY / Adenosylhomocysteinase Tested Reactivity Hu, Ms Tested Application WB Host Rabbit Clonality Polyclonal Isotype IgG Target Name AHCY / Adenosylhomocysteinase Antigen Species Human Immunogen Recombinant Protein of Human AHCY / Adenosylhomocysteinase. Conjugation Un-conjugated Alternate Names adoHcyase; AdoHcyase; SAHH; Adenosylhomocysteinase; EC 3.3.1.1; S-adenosyl-L-homocysteine hydrolase Application Instructions Application table Application Dilution WB 1:500 - 1:2000 Application Note * The dilutions indicate recommended starting dilutions and the optimal dilutions or concentrations should be determined by the scientist. Positive Control SW480 Calculated Mw 48 kDa Properties Form Liquid Purification Affinity purification with immunogen. Buffer PBS (pH 7.3), 0.02% Sodium azide and 50% Glycerol. Preservative 0.02% Sodium azide Stabilizer 50% Glycerol Storage instruction For continuous use, store undiluted antibody at 2-8°C for up to a week. For long-term storage, aliquot and store at -20°C. Storage in frost free freezers is not recommended. Avoid repeated freeze/thaw cycles. Suggest spin the vial prior to opening. The antibody solution should be gently mixed before use. Note For laboratory research only, not for drug, diagnostic or other use. www.arigobio.com 1/2 Bioinformation Gene Symbol AHCY Gene Full Name adenosylhomocysteinase Background S-adenosylhomocysteine hydrolase belongs to the adenosylhomocysteinase family. It catalyzes the reversible hydrolysis of S-adenosylhomocysteine (AdoHcy) to adenosine (Ado) and L-homocysteine (Hcy). Thus, it regulates the intracellular S-adenosylhomocysteine (SAH) concentration thought to be important for transmethylation reactions.
    [Show full text]
  • Biochemistry I Enzymes
    BIOCHEMISTRY I 3rd. Stage Lec. ENZYMES Biomedical Importance: Enzymes, which catalyze the biochemical reactions, are essential for life. They participate in the breakdown of nutrients to supply energy and chemical building blocks; the assembly of those building blocks into proteins, DNA, membranes, cells, and tissues; and the harnessing of energy to power cell motility, neural function, and muscle contraction. The vast majority of enzymes are proteins. Notable exceptions include ribosomal RNAs and a handful of RNA molecules imbued with endonuclease or nucleotide ligase activity known collectively as ribozymes. The ability to detect and to quantify the activity of specific enzymes in blood, other tissue fluids, or cell extracts provides information that complements the physician’s ability to diagnose and predict the prognosis of many diseases. Further medical applications include changes in the quantity or in the catalytic activity of key enzymes that can result from genetic defects, nutritional deficits, tissue damage, toxins, or infection by viral or bacterial pathogens (eg, Vibrio cholerae). Medical scientists address imbalances in enzyme activity by using pharmacologic agents to inhibit specific enzymes and are investigating gene therapy as a means to remedy deficits in enzyme level or function. In addition to serving as the catalysts for all metabolic processes, their impressive catalytic activity, substrate specificity, and stereospecificity enable enzymes to fulfill key roles in additional processes related to human health and well-being. Proteases and amylases augment the capacity of detergents to remove dirt and stains, and enzymes play important roles in producing or enhancing the nutrient value of food products for both humans and animals.
    [Show full text]
  • GPX4 at the Crossroads of Lipid Homeostasis and Ferroptosis Giovanni C
    REVIEW GPX4 www.proteomics-journal.com GPX4 at the Crossroads of Lipid Homeostasis and Ferroptosis Giovanni C. Forcina and Scott J. Dixon* formation of toxic radicals (e.g., R-O•).[5] Oxygen is necessary for aerobic metabolism but can cause the harmful The eight mammalian GPX proteins fall oxidation of lipids and other macromolecules. Oxidation of cholesterol and into three clades based on amino acid phospholipids containing polyunsaturated fatty acyl chains can lead to lipid sequence similarity: GPX1 and GPX2; peroxidation, membrane damage, and cell death. Lipid hydroperoxides are key GPX3, GPX5, and GPX6; and GPX4, GPX7, and GPX8.[6] GPX1–4 and 6 (in intermediates in the process of lipid peroxidation. The lipid hydroperoxidase humans) are selenoproteins that contain glutathione peroxidase 4 (GPX4) converts lipid hydroperoxides to lipid an essential selenocysteine in the enzyme + alcohols, and this process prevents the iron (Fe2 )-dependent formation of active site, while GPX5, 6 (in mouse and toxic lipid reactive oxygen species (ROS). Inhibition of GPX4 function leads to rats), 7, and 8 use an active site cysteine lipid peroxidation and can result in the induction of ferroptosis, an instead. Unlike other family members, GPX4 (PHGPx) can act as a phospholipid iron-dependent, non-apoptotic form of cell death. This review describes the hydroperoxidase to reduce lipid perox- formation of reactive lipid species, the function of GPX4 in preventing ides to lipid alcohols.[7,8] Thus,GPX4ac- oxidative lipid damage, and the link between GPX4 dysfunction, lipid tivity is essential to maintain lipid home- oxidation, and the induction of ferroptosis. ostasis in the cell, prevent the accumula- tion of toxic lipid ROS and thereby block the onset of an oxidative, iron-dependent, non-apoptotic mode of cell death termed 1.
    [Show full text]
  • Prioritization of Metabolic Genes As Novel Therapeutic Targets in Estrogen-Receptor Negative Breast Tumors Using Multi-Omics Data and Text Mining
    www.oncotarget.com Oncotarget, 2019, Vol. 10, (No. 39), pp: 3894-3909 Research Paper Prioritization of metabolic genes as novel therapeutic targets in estrogen-receptor negative breast tumors using multi-omics data and text mining Dinesh Kumar Barupal1,*, Bei Gao1,*, Jan Budczies2, Brett S. Phinney4, Bertrand Perroud4, Carsten Denkert2,3 and Oliver Fiehn1 1West Coast Metabolomics Center, University of California, Davis, CA, USA 2Institute of Pathology, Charité University Hospital, Berlin, Germany 3German Institute of Pathology, Philipps-University Marburg, Marburg, Germany 4UC Davis Genome Center, University of California, Davis, CA, USA *Co-first authors and contributed equally to this work Correspondence to: Oliver Fiehn, email: [email protected] Keywords: set-enrichment; ChemRICH; multi-omics; metabolic networks; candidate gene prioritization Received: March 12, 2019 Accepted: May 13, 2019 Published: June 11, 2019 Copyright: Barupal et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Estrogen-receptor negative (ERneg) breast cancer is an aggressive breast cancer subtype in the need for new therapeutic options. We have analyzed metabolomics, proteomics and transcriptomics data for a cohort of 276 breast tumors (MetaCancer study) and nine public transcriptomics datasets using univariate statistics, meta- analysis, Reactome pathway analysis, biochemical network mapping and text mining of metabolic genes. In the MetaCancer cohort, a total of 29% metabolites, 21% proteins and 33% transcripts were significantly different (raw p <0.05) between ERneg and ERpos breast tumors.
    [Show full text]
  • The Microbiota-Produced N-Formyl Peptide Fmlf Promotes Obesity-Induced Glucose
    Page 1 of 230 Diabetes Title: The microbiota-produced N-formyl peptide fMLF promotes obesity-induced glucose intolerance Joshua Wollam1, Matthew Riopel1, Yong-Jiang Xu1,2, Andrew M. F. Johnson1, Jachelle M. Ofrecio1, Wei Ying1, Dalila El Ouarrat1, Luisa S. Chan3, Andrew W. Han3, Nadir A. Mahmood3, Caitlin N. Ryan3, Yun Sok Lee1, Jeramie D. Watrous1,2, Mahendra D. Chordia4, Dongfeng Pan4, Mohit Jain1,2, Jerrold M. Olefsky1 * Affiliations: 1 Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California, USA. 2 Department of Pharmacology, University of California, San Diego, La Jolla, California, USA. 3 Second Genome, Inc., South San Francisco, California, USA. 4 Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA. * Correspondence to: 858-534-2230, [email protected] Word Count: 4749 Figures: 6 Supplemental Figures: 11 Supplemental Tables: 5 1 Diabetes Publish Ahead of Print, published online April 22, 2019 Diabetes Page 2 of 230 ABSTRACT The composition of the gastrointestinal (GI) microbiota and associated metabolites changes dramatically with diet and the development of obesity. Although many correlations have been described, specific mechanistic links between these changes and glucose homeostasis remain to be defined. Here we show that blood and intestinal levels of the microbiota-produced N-formyl peptide, formyl-methionyl-leucyl-phenylalanine (fMLF), are elevated in high fat diet (HFD)- induced obese mice. Genetic or pharmacological inhibition of the N-formyl peptide receptor Fpr1 leads to increased insulin levels and improved glucose tolerance, dependent upon glucagon- like peptide-1 (GLP-1). Obese Fpr1-knockout (Fpr1-KO) mice also display an altered microbiome, exemplifying the dynamic relationship between host metabolism and microbiota.
    [Show full text]
  • Differential Effects on Growth, Homocysteine, and Related
    (CANCER RESEARCH 49, 324-330, January 15, 1989] Differential Effects on Growth, Homocysteine, and Related Compounds of Two Inhibitors of 5-AdenosyIhomocysteinc Catabolism, 3-Deazaadenosine, and 3-Deazaaristeromycin, in C3H/10T1/2 Cells1 Rune Djurhuus,2 Asbjorn M. Svardal,3 and Per M. Ueland Clinical Pharmacology Unit, Department of Pharmacology and Toxicology, University of Bergen, Norway ABSTRACT logues were shown to interfere with the function of AdoHcy hydrolase, and this enzyme was used as a target for design of The growth of nontransformed (Cl 8) and malignant (C) 16) C3H/ biological active compounds (3, 4). c3Ari is a result of such Kll 1/2 mouse embryo fibroblasts was inhibited by 3-deazaadenosine (c'Ailii) (I.I)«,= 195 MMfor Cl 8 and 30 MMfor CI 16 cells) and 3- rational design (5). deazaaristeromycin (c'Ari) (LDM about 36 MMfor Cl 8 and 9 MMfor Cl Adenosine analogues may function as competitive inhibitor, 16 cells). Both compounds inhibited in a dose-dependent manner .V- substrate, or inactivator (irreversible inhibitor) of AdoHcy hy adenosylhomocysteine (AdoHcy) catabolisnt and homocysteine produc drolase (2). In principle, the metabolic derangements induced tion, measured as homocysteine egress, and c'Ari was most potent in this by agents interacting with this enzyme can be divided into two respect. c'Ad»gave rise to its congener, 3-deazaadenosylhomocysteine types: (c3AdoHcy). Addition of homocysteine thiolactone (Hcy-tl) to the medium (a) Inhibition of AdoHcy catabolism results in accumulation enhanced AdoHcy (and c3AdoHcy) accumulation but did not affect the of large amount of this metabolite, and nucleosides serving as cell growth at concentrations of inhibitor less than 10 MM.
    [Show full text]
  • The C718T Polymorphism in the 3″-Untranslated
    Hypertension Research (2012) 35, 507–512 & 2012 The Japanese Society of Hypertension All rights reserved 0916-9636/12 www.nature.com/hr ORIGINAL ARTICLE The C718T polymorphism in the 3¢-untranslated region of glutathione peroxidase-4 gene is a predictor of cerebral stroke in patients with essential hypertension Alexey V Polonikov1, Ekaterina K Vialykh2, Mikhail I Churnosov3, Thomas Illig4, Maxim B Freidin5, Oksana V Vasil¢eva1, Olga Yu Bushueva1, Valentina N Ryzhaeva1, Irina V Bulgakova1 and Maria A Solodilova1 In the present study we have investigated the association of three single nucleotide polymorphisms in glutathione peroxidase (GPx) genes GPX1 rs1050450 (P198L), GPX3 rs2070593 (G930A) and GPX4 rs713041 (T718C) with the risk of cerebral stroke (CS) in patients with essential hypertension (EH). A total of 667 unrelated EH patients of Russian origin, including 306 hypertensives (the EH–CS group) who suffered from CS and 361 people (the EH–CS group) who did not have cerebrovascular accidents, were enrolled in the study. The variant allele 718C of the GPX4 gene was found to be significantly associated with an increased risk of CS in hypertensive patients (odds ratio (OR) 1.53, 95% confidence interval (CI) 1.23–1.90, Padj¼0.0003). The prevalence of the 718TC and 718CC genotypes of the GPX4 gene was higher in the EH–CS group than the EH-alone group (OR¼2.12, 95%CI 1.42–3.16, Padj¼0.0018). The association of the variant GPX4 genotypes with the increased risk of CS in hypertensives remained statistically significant after adjusting for confounding variables such as sex, body mass index (BMI), blood pressure and antihypertensive medication use (OR¼2.18, 95%CI 1.46–3.27, P¼0.0015).
    [Show full text]
  • Programmed Cell-Death by Ferroptosis: Antioxidants As Mitigators
    International Journal of Molecular Sciences Review Programmed Cell-Death by Ferroptosis: Antioxidants as Mitigators Naroa Kajarabille 1 and Gladys O. Latunde-Dada 2,* 1 Nutrition and Obesity Group, Department of Nutrition and Food Sciences, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain; [email protected] 2 King’s College London, Department of Nutritional Sciences, Faculty of Life Sciences and Medicine, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK * Correspondence: [email protected] Received: 9 September 2019; Accepted: 2 October 2019; Published: 8 October 2019 Abstract: Iron, the fourth most abundant element in the Earth’s crust, is vital in living organisms because of its diverse ligand-binding and electron-transfer properties. This ability of iron in the redox cycle as a ferrous ion enables it to react with H2O2, in the Fenton reaction, to produce a hydroxyl radical ( OH)—one of the reactive oxygen species (ROS) that cause deleterious oxidative damage • to DNA, proteins, and membrane lipids. Ferroptosis is a non-apoptotic regulated cell death that is dependent on iron and reactive oxygen species (ROS) and is characterized by lipid peroxidation. It is triggered when the endogenous antioxidant status of the cell is compromised, leading to lipid ROS accumulation that is toxic and damaging to the membrane structure. Consequently, oxidative stress and the antioxidant levels of the cells are important modulators of lipid peroxidation that induce this novel form of cell death. Remedies capable of averting iron-dependent lipid peroxidation, therefore, are lipophilic antioxidants, including vitamin E, ferrostatin-1 (Fer-1), liproxstatin-1 (Lip-1) and possibly potent bioactive polyphenols.
    [Show full text]